Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 70
1.
Cereb Cortex ; 32(2): 286-297, 2022 01 10.
Article En | MEDLINE | ID: mdl-34223882

Folic acid (FA) has been reported to inhibit astrocyte apoptosis and improve aging-induced disorders; however, its role in telomere attrition remains unclear. In present study, 4-month-old senescence-accelerated mouse prone 8 (SAMP8) mice were assigned to four treatment groups for the in vivo experiment: FA-deficient diet (FA-D) group, FA-normal diet (FA-N) group, low FA-supplemented diet (FA-L) group, and high FA-supplemented diet (FA-H) group. These mice were euthanized when 10 months old. There was also a young SAMP8 (4 months old) control group (Con-Y) fed with FA-normal diet. In in vitro study, primary cultures of astrocytes from hippocampus and cerebral cortex were incubated for five generations with various concentrations of FA (0-40 µM) and were assigned to five groups: FA 0 µM (generation 5), FA 10 µM (generation 5), FA 20 µM (generation 5), FA 40 µM (generation 5), and FA 10 µM (generation 1). The results showed that FA supplementation inhibited aging-induced astrocytosis, astrocyte apoptosis, neurodegeneration, and prevented telomere attrition in hippocampus and cortex of SAMP8 mice. FA supplementation also decreased apoptosis and telomere attrition, and increased telomerase activity, in primary cultures of astrocytes. These results showed that it may be one of the mechanisms that FA inhibiting aging-induced apoptosis of astrocyte by alleviating telomere attrition.


Astrocytes , Folic Acid , Aging , Animals , Apoptosis , Folic Acid/pharmacology , Mice , Telomere
2.
Mol Neurobiol ; 59(1): 590-602, 2022 Jan.
Article En | MEDLINE | ID: mdl-34741234

DNA oxidative damage can cause telomere attrition or dysfunction that triggers cell senescence and apoptosis. The hypothesis of this study is that folic acid decreases apoptosis in neural stem cells (NSCs) by preventing oxidative stress-induced telomere attrition. Primary cultures of NSCs were incubated for 9 days with various concentrations of folic acid (0-40 µM) and then incubated for 24 h with a combination of folic acid and an oxidant (100-µM hydrogen peroxide, H2O2), antioxidant (10-mM N-acetyl-L-cysteine, NAC), or vehicle. Intracellular folate concentration, apoptosis rate, cell proliferative capacity, telomere length, telomeric DNA oxidative damage, telomerase activity, intracellular reactive oxygen species (ROS) levels, cellular oxidative damage, and intracellular antioxidant enzyme activities were determined. The results showed that folic acid deficiency in NSCs decreased intracellular folate concentration, cell proliferation, telomere length, and telomerase activity but increased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. In contrast, folic acid supplementation dose-dependently increased intracellular folate concentration, cell proliferative capacity, telomere length, and telomerase activity but decreased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. Exposure to H2O2 aggravated telomere attrition and oxidative damage, whereas NAC alleviated the latter. High doses of folic acid prevented telomere attrition and telomeric DNA oxidative damage by H2O2. In conclusion, inhibition of telomeric DNA oxidative damage and telomere attrition in NSCs may be potential mechanisms of inhibiting NSC apoptosis by folic acid.


Antioxidants/pharmacology , Apoptosis/drug effects , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Oxidative Stress/drug effects , Telomere/drug effects , Animals , Cell Proliferation/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Neural Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Telomere/metabolism
3.
Int J Mol Sci ; 21(1)2019 Dec 20.
Article En | MEDLINE | ID: mdl-31861819

Astrocytes are the most widely distributed cells in the brain, and astrocyte apoptosis may play an important role in the pathogenesis of neurodegenerative diseases. Folate is required for the normal development of the nervous system, but its effect on astrocyte apoptosis is unclear. In this study, we hypothesized that folic acid (the therapeutic form of folate) decreases astrocyte apoptosis by preventing oxidative stress-induced telomere attrition. Primary cultures of astrocytes were incubated for 12 days with various concentrations of folic acid (0-40 µmol/L), then cell proliferation, apoptosis, intracellular folate concentration, intracellular homocysteine (Hcy) concentration, intracellular reactive oxygen species (ROS) levels, telomeric DNA oxidative damage, and telomere length were determined. The results showed that folic acid deficiency decreased intracellular folate, cell proliferation, and telomere length, whereas it increased Hcy concentration, ROS levels, telomeric DNA oxidative damage, and apoptosis. In contrast, folic acid dose-dependently increased intracellular folate, cell proliferation, and telomere length but it decreased Hcy concentration, ROS levels, telomeric DNA oxidative damage, and apoptosis. In conclusion, folic acid inhibited apoptosis in astrocytes. The underlying mechanism for this protective effect may be that folic acid decreased oxidative stress and thereby prevented telomeric DNA oxidative damage and telomere attrition.


Apoptosis/drug effects , Astrocytes/drug effects , Folic Acid/pharmacology , Oxidative Stress/drug effects , Vitamin B Complex/pharmacology , Animals , Antioxidants/pharmacology , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Rats, Sprague-Dawley , Telomere/drug effects , Telomere/metabolism
4.
Aging (Albany NY) ; 11(22): 10356-10373, 2019 11 22.
Article En | MEDLINE | ID: mdl-31757935

The occurrence of telomere attrition in brain may cause senescence and death of neurons, leading to cognitive decline. Folic acid (FA) has been reported to improve cognitive performance in mild cognitive impairment; however, its association with telomere remains unclear. The study aimed to investigate if alleviation of telomere attrition by FA supplementation could act as a potential mechanism to delay age-related cognitive decline in senescence-accelerated mouse prone 8 (SAMP8). Aged SAMP8 mice were assigned to four treatment groups: FAdeficient diet (FA-D) group, FA-normal diet (FA-N) group, low FA-supplemented diet (FA-L) group and high FAsupplemented diet (FA-H) group. There was also an age-matched senescence-accelerated mouse resistant 1 (SAMR1) control group (Con-R), and a young SAMP8 control group (Con-Y). The results demonstrated that FA supplementation delayed age-related cognitive decline and neurodegeneration in SAMP8 mice. Importantly, this effect could be attributed to the alleviated telomere attrition, which might be interpreted by the decreased levels of reactive oxygen species. Additionally, improved telomere integrity stimulated mitochondrial function via telomere-p53-mithondria pathway, consequently delayed neuronal degeneration. In conclusion, we demonstrate that FA supplementation delays age-related neurodegeneration and cognitive decline in SAMP8 mice, in which alleviated telomere attrition could serve as one influential factor in the process.


Aging/drug effects , Cognitive Dysfunction , Dietary Supplements , Folic Acid/pharmacology , Telomere Shortening/drug effects , Animals , Brain/drug effects , Brain/pathology , Male , Mice , Nerve Degeneration/pathology
5.
Cereb Cortex ; 29(8): 3390-3397, 2019 07 22.
Article En | MEDLINE | ID: mdl-30137237

Maternal folic acid supplementation during pregnancy is associated with improved cognitive performances in offspring. However, the effect of supplementation on offspring's neurogenesis and synaptogenesis is unknown, and whether supplementation should be continued throughout pregnancy is controversial. In present study, 3 groups of female rats were fed a folate-normal diet, folate-deficient diet, or folate-supplemented diet from 1 week before mating until the end of pregnancy. A fourth group fed folate-normal diet from 1 week before mating until mating, then fed folate-supplemented diet for 10 consecutive days, then fed folate-normal diet until the end of pregnancy. Offspring were sacrificed on postnatal day 0 for measurement of neurogenesis and synaptogenesis by immunofluorescence and western blot. Additionally neural stem cells (NSCs) were cultured from offspring's hippocampus for immunocytochemical measurement of their rates of proliferation and neuronal differentiation. The results demonstrated that maternal folic acid supplementation stimulated hippocampal neurogenesis by increasing proliferation and neuronal differentiation of NSCs, and also enhanced synaptogenesis in cerebral cortex of neonatal offspring. Hippocampal neurogenesis was stimulated more when supplementation was continued throughout pregnancy instead of being limited to the periconceptional period. In conclusion, maternal folic acid supplementation, especially if continued throughout pregnancy, improves neurogenesis and synaptogenesis in neonatal offspring.


Cell Proliferation/drug effects , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Synapses/drug effects , Vitamin B Complex/pharmacology , Animals , Animals, Newborn , Blotting, Western , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival , Duration of Therapy , Female , Hippocampus/cytology , Hippocampus/drug effects , Pregnancy , Primary Cell Culture , Random Allocation , Rats , Synaptophysin/drug effects , Synaptophysin/metabolism
6.
Nutrients ; 10(3)2018 Mar 01.
Article En | MEDLINE | ID: mdl-29494536

Periconceptional maternal folate levels may alter DNA methylation patterns and health outcomes in offspring. We hypothesized that maternal folic acid supplementation alters fetal neural development through DNA methylation in the fetal brain. Twenty-eight rats were randomly assigned to four groups: three groups of the female rats were fed folate-normal, folate-deficient or folate-supplemented diets from seven days before mating to delivery. In another group, folic acid supplementation diet short-period group was fed a folate-normal diet, except for 10 days (begin mating) when this group was fed a folate-supplemented diet. After delivery, the diets were changed to folate-normal diet for all four groups. The cliff avoidance and forelimb grip tests were used to assess sensory motor function of rat offspring. The results indicate that maternal folic acid supplementation improved the early development of sensory-motor function in offspring. Maternal folic acid supplementation increased the methylation potential, global DNA methylation (5-mC) and DNA methyltransferase expression and activity in the brains of the offspring. In conclusion, maternal folic acid supplementation increases DNA methylation pattern in offspring brain and improves the early development of sensory-motor function.


DNA Methylation/drug effects , Dietary Supplements , Folic Acid/administration & dosage , Animals , Behavior, Animal , Female , Litter Size , Male , Pregnancy , Prenatal Exposure Delayed Effects , Prenatal Nutritional Physiological Phenomena , Random Allocation , Rats , Resistance Training
7.
Mol Neurobiol ; 55(3): 2676-2684, 2018 03.
Article En | MEDLINE | ID: mdl-28421540

Maternal folate status during pregnancy may influence central nervous system (CNS) development in offspring. However, the recommended intakes of folic acid for women of childbearing age differ among countries and there is still no consensus about whether folic acid should be supplemented continuously throughout pregnancy. We hypothesized that folic acid supplementation may be more beneficial for offspring's neurobehavioral development if prolonged throughout pregnancy instead of being limited to the periconceptional period. In this study, three groups of the female rats were fed folate-normal, folate-deficient, or folate-supplemented diets throughout pregnancy. In another group, the female rats were fed folate-supplemented diet from mating for 10 consecutive days and then fed folate-normal diet for remainder days of pregnancy. The results showed that maternal folate deficiency increased plasma homocysteine (Hcy) concentration in dams, delayed early sensory-motor reflex development, impaired spatial learning and memory ability, and caused ultrastructural damages in the hippocampus of offspring. Maternal folic acid supplementation would be more effective on improving early sensory-motor reflex development and spatial learning and memory ability in offspring if prolonged throughout pregnancy instead of being limited to the periconceptional period. In conclusion, prolonged maternal folic acid supplementation throughout pregnancy would be more effective in neurobehavioral development of offspring in rats.


Dietary Supplements , Folic Acid/administration & dosage , Learning/drug effects , Pregnancy/drug effects , Psychomotor Performance/drug effects , Reflex, Righting/drug effects , Age Factors , Animals , Animals, Newborn , Female , Hippocampus/drug effects , Hippocampus/physiology , Hippocampus/ultrastructure , Learning/physiology , Male , Maternal Health , Pregnancy/physiology , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Reflex, Righting/physiology
8.
Mol Neurobiol ; 53(9): 6548-6556, 2016 11.
Article En | MEDLINE | ID: mdl-26627706

Dementia has emerged as a major societal issue because of the worldwide aging population and the absence of any effective treatment. DNA methylation is an epigenetic mechanism that evidently plays a role in Alzheimer's disease (AD). Folate acts through one-carbon metabolism to support the methylation of multiple substrates including DNA. We aimed to test the hypothesis that folic acid supplementation alters DNA methylation profiles in AD models. Mouse Neuro-2a cells expressing human APP695 (N2a-APP cells) were incubated with folic acid (2.8-20 µmol/L). AD transgenic mice were fed either folate-deficient or control diets and gavaged daily with water or folic acid (600 µg/kg). Gene methylation profiles were determined by methylated DNA immunoprecipitation-DNA microarray (MeDIP-chip). Differentially methylated regions (DMRs) were determined by Quantitative Differentially Methylated Regions analysis, and differentially methylated genes (DMGs) carrying at least three DMRs were selected for pathway analysis. Folic acid up-regulated DNA methylation levels in N2a-APP cells and AD transgenic mouse brains. Functional network analysis of folic acid-induced DMGs in these AD models revealed subnetworks composed of 24 focus genes in the janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway and 12 focus genes in the long-term depression (LTD) signaling pathway. In conclusion, these results revealed a role for folic acid in the JAK-STAT and LTD signaling pathways which may be relevant to AD pathogenesis. This novel finding may stimulate reinvestigation of folic acid supplementation as a prophylactic or therapeutic treatment for AD.


Alzheimer Disease/genetics , DNA Methylation/drug effects , Folic Acid/pharmacology , Janus Kinases/metabolism , Long-Term Synaptic Depression/drug effects , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Humans , Male , Mice, Transgenic , Reproducibility of Results , Signal Transduction/genetics
9.
Eur J Nutr ; 55(5): 1849-62, 2016 Aug.
Article En | MEDLINE | ID: mdl-26224648

PURPOSE: Alzheimer's disease (AD) is a highly prevalent type of dementia. The epigenetic mechanism of gene methylation provides a putative link between nutrition, one-carbon metabolism, and disease progression because folate deficiency may cause hypomethylation of promoter regions in AD-relevant genes. We hypothesized that folic acid supplementation may protect neuron cells from amyloid ß (Aß) oligomer-induced toxicity by modulating DNA methylation of APP and PS1 in AD models. METHODS: Primary hippocampal neuronal cells and hippocampal HT-22 cells were incubated for 24 h with a combination of folic acid and either Aß oligomers or vehicle and were then incubated for 72 h with various concentrations of folic acid. AD transgenic mice were fed either folate-deficient or control diets and gavaged daily with various doses of folic acid (0 or 600 µg/kg). DNA methyltransferase (DNMT) activity, cell viability, methylation potential of cells, APP and PS1 expression, and the methylation of the respective promoters were determined. RESULTS: Aß oligomers lowered DNMT activity, increased PS1 and APP expression, and decreased cell viability. Folic acid dose-dependently stimulated methylation potential and DNMT activity, altered PS1 and APP promoter methylation, decreased PS1 and APP expression, and partially preserved cell viability. Folic acid increased PS1 and APP promoter methylation in AD transgenic mice. CONCLUSION: These results suggest a mechanism by which folic acid may prevent Aß oligomer-induced neuronal toxicity.


Amyloid beta-Peptides/metabolism , DNA Methylation/drug effects , Folic Acid/pharmacology , Neurons/drug effects , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Animals , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Folic Acid/blood , Folic Acid Deficiency/blood , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Promoter Regions, Genetic
10.
Int J Mol Sci ; 16(10): 25002-13, 2015 Oct 20.
Article En | MEDLINE | ID: mdl-26492244

Alzheimer's disease (AD) is a common neurodegenerative disease resulting in progressive dementia, and is a principal cause of dementia among older adults. Folate acts through one-carbon metabolism to support the methylation of multiple substrates. We hypothesized that folic acid supplementation modulates DNA methyltransferase (DNMT) activity and may alter amyloid ß-peptide (Aß) production in AD. Mouse Neuro-2a cells expressing human APP695 were incubated with folic acid (2.8-40 µmol/L), and with or without zebularine (the DNMT inhibitor). DNMT activity, cell viability, Aß and DNMTs expression were then examined. The results showed that folic acid stimulated DNMT gene and protein expression, and DNMT activity. Furthermore, folic acid decreased Aß protein production, whereas inhibition of DNMT activity by zebularine increased Aß production. The results indicate that folic acid induces methylation potential-dependent DNMT enzymes, thereby attenuating Aß production.


Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Folic Acid/therapeutic use , Alzheimer Disease/enzymology , Animals , Cell Line, Tumor , DNA Methylation/drug effects , DNA Modification Methylases/metabolism , Enzyme Activation/drug effects , Folic Acid/pharmacology , Humans , Mice
11.
J Nutr Biochem ; 26(8): 883-91, 2015 Aug.
Article En | MEDLINE | ID: mdl-25959374

Alzheimer's disease (AD) is associated with malnutrition, altered one-carbon metabolism and increased hippocampal amyloid-ß peptide (Aß) accumulation. Aberrant DNA methylation may be an epigenetic mechanism that underlies AD pathogenesis. We hypothesized that folic acid acts through an epigenetic gene silencing mechanism to lower Aß levels in the APP/PS1 transgenic mouse model of AD. APP/PS1 mice were fed either folate-deficient or control diets and gavaged daily with 120 µg/kg folic acid, 13.3mg/kg S-adenosylmethionine (SAM) or both. Examination of the mice after 60 days of treatment showed that serum folate concentration increased with intake of folic acid but not SAM. Folate deficiency lowered endogenous SAM concentration, whereas neither intervention altered S-adenosylhomocysteine concentration. DNA methyltransferase (DNMT) activity increased with intake of folic acid raised DNMT activity in folate-deficient mice. DNA methylation rate was stimulated by folic acid in the amyloid precursor protein (APP) promoter and in the presenilin 1 (PS1) promoter. Folate deficiency elevated hippocampal APP, PS1 and Aß protein levels, and these rises were prevented by folic acid. In conclusion, these findings are consistent with a mechanism in which folic acid increases methylation potential and DNMT activity, modifies DNA methylation and ultimately decreases APP, PS1 and Aß protein levels.


Amyloid beta-Peptides/metabolism , Folic Acid/administration & dosage , Amino Acid Sequence , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Animals , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Folic Acid/blood , Folic Acid Deficiency/blood , Gene Silencing , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Presenilin-1/genetics , Presenilin-1/metabolism , Promoter Regions, Genetic , S-Adenosylhomocysteine/blood , S-Adenosylmethionine/blood , Sequence Analysis, DNA
12.
Blood Coagul Fibrinolysis ; 26(4): 436-42, 2015 Jun.
Article En | MEDLINE | ID: mdl-25730478

The microcirculation during sepsis fails due to capillary plugging involving microthrombosis. We demonstrated that intravenous injection of ascorbate reduces this plugging, but the mechanism of this beneficial effect remains unclear. We hypothesize that ascorbate inhibits the release of the antifibrinolytic plasminogen activator inhibitor-1 (PAI-1) from endothelial cells and platelets during sepsis. Microvascular endothelial cells and platelets were isolated from mice. Cells were cultured and stimulated with lipopolysaccharide (LPS), tumor necrosis factor alpha (TNFα), or thrombin (agents of sepsis), with/without ascorbate for 1-24 h. PAI-1 mRNA was determined by quantitative PCR. PAI-1 protein release into the culture medium was measured by ELISA. In platelets, PAI-1 release was measured after LPS, TNFα, or thrombin stimulation, with/without ascorbate. In endothelial cells, LPS and TNFα increased PAI-1 mRNA after 6-24 h, but no increase in PAI-1 release was observed; ascorbate did not affect these responses. In platelets, thrombin, but not LPS or TNFα, increased PAI-1 release; ascorbate inhibited this increase at low extracellular pH. In unstimulated endothelial cells and platelets, PAI-1 is released into the extracellular space. Thrombin increases this release from platelets; ascorbate inhibits it pH-dependently. The data suggest that ascorbate promotes fibrinolysis in the microvasculature under acidotic conditions in sepsis.


Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Blood Platelets/drug effects , Endothelial Cells/drug effects , Plasminogen Activator Inhibitor 1/genetics , Sepsis/drug therapy , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/immunology , RNA, Messenger/genetics , Sepsis/genetics , Sepsis/immunology
13.
J Surg Res ; 191(2): 432-40, 2014 Oct.
Article En | MEDLINE | ID: mdl-24814200

BACKGROUND: Sepsis, a potential risk associated with surgery, leads to a systemic inflammatory response including the plugging of capillary beds. This plugging may precipitate organ failure and subsequent death. We have shown that capillary plugging can be reversed rapidly within 1 h by intravenous injection of ascorbate in mouse skeletal muscle. It is unknown whether, in parallel with this effect, ascorbate negatively affects the protective responses to sepsis involving the fibrinolytic and immune systems. We hypothesized that treatment with ascorbate for 1 h does not alter bacterial content, plasminogen activator inhibitor 1 (PAI-1), and neutrophil infiltration in lung, kidney, spleen, and liver (organs with high immune response) of septic mice. MATERIALS AND METHODS: Sepsis was induced by feces injection into the peritoneum. Mice were injected intravenously with ascorbate at 6 h (10 mg/kg), and samples of peritoneal fluid, arterial blood, and organs collected at 7 h were subjected to analyses of bacterial content, PAI-1 messenger RNA and enzymatic activity, and myeloperoxidase (MPO) (a measure of neutrophil infiltration). RESULTS: Sepsis increased bacterial content in all fluids and organs and increased PAI-1 messenger RNA and enzymatic activity in the lung and liver. Sepsis increased the myeloperoxidase level in the lung and liver, and lowered it in the spleen. Except for decreasing the bacterial content in blood, these responses to sepsis were not altered by ascorbate. CONCLUSIONS: The rapid effect of ascorbate against capillary plugging in the septic mouse skeletal muscle is not accompanied by alterations in PAI-1 or myeloperoxidase responses in the organs with high immune response.


Ascorbic Acid/pharmacology , Bacteria/isolation & purification , Peroxidase/metabolism , Plasminogen Activator Inhibitor 1/genetics , Sepsis/drug therapy , Animals , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , RNA, Messenger/analysis , Sepsis/immunology , Sepsis/microbiology
14.
Blood Coagul Fibrinolysis ; 25(7): 745-53, 2014 Oct.
Article En | MEDLINE | ID: mdl-24824492

Plugging of the capillary bed in tissues correlates with organ failure during sepsis. In septic mouse skeletal muscle, we showed that blood in capillaries becomes hypercoagulable and that ascorbate injection inhibits capillary plugging. In the present study, we hypothesized that ascorbate promotes fibrinolysis, reversing this plugging. Sepsis in mice was induced by fecal injection into peritoneum. Mice were injected intravenously with a bolus of streptokinase (fibrinolytic agent) or ascorbate at 5-6 h. Both agents reversed capillary plugging in muscle at 7 h. Sepsis increased mRNA expression of urokinase plasminogen activator (u-PA) (profibrinolytic) and plasminogen activator inhibitor 1 (PAI-1) (antifibrinolytic) in muscle and liver homogenates at 7 h. Ascorbate did not affect u-PA mRNA in either tissue, but it inhibited PAI-1 mRNA in muscle, suggesting enhanced fibrinolysis in this tissue. However, ascorbate did not affect increased PAI-1 mRNA in the liver (dominant source of soluble PAI-1 in systemic blood). Consistently, ascorbate affected neither elevated PAI-1 protein/enzymatic activity in septic liver nor lowered plasmin antiplasmin level in septic blood. Furthermore, hypocoagulability of septic blood revealed by thrombelastography and thrombin-induced PAI-1 release from isolated platelets (ex-vivo model of sepsis) were not affected by ascorbate. Based on the PAI-1 protein data, the present study does not support the hypothesis that ascorbate promotes fibrinolysis in sepsis.


Ascorbic Acid/pharmacology , Fibrinolysis/drug effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Sepsis/blood , Animals , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/pathology , Serpin E2/genetics , Serpin E2/metabolism , Thrombelastography/methods , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
15.
J Nutr Biochem ; 25(4): 496-502, 2014 Apr.
Article En | MEDLINE | ID: mdl-24629913

Proliferation of neural stem cells (NSCs) is required for development and repair in the nervous system. NSC amplification in vitro is a necessary step towards using NSC transplantation therapy to treat neurodegenerative diseases. Folic acid (FA) has been shown to act through DNA methyltransferase to stimulate NSC proliferation. To elucidate the underlying mechanism, the effect of FA on the methylation profiles in neonatal rat NSCs was assessed by methylated DNA immunoprecipitation (MeDIP) and methylated DNA immunoprecipitation-DNA microarray (MeDIP-Chip). Differentially methylated regions (DMRs) were determined by quantitative differentially methylated regions analysis, and genes carrying at least three DMRs were selected for pathway analysis. Gene network analysis revealed links with steroid biosynthesis, fatty acid elongation and the PI3K/Akt/CREB, neuroactive ligand-receptor interaction, Jak-STAT and MAPK signaling pathways. Moreover, Akt3 acted as a hub in the network, in which 14 differentially methylated genes converged to the PI3K/Akt/CREB signaling pathway. These findings indicate that FA stimulates NSC proliferation by modifying DNA methylation levels in the PI3K/Akt/CREB pathway.


Cyclic AMP Response Element-Binding Protein/metabolism , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , DNA Methylation/drug effects , DNA Modification Methylases/metabolism , Dose-Response Relationship, Drug , Female , Gene Regulatory Networks/drug effects , Methylation , Neural Stem Cells/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects
16.
J Nutr Biochem ; 24(11): 1817-22, 2013 Nov.
Article En | MEDLINE | ID: mdl-23850087

Folic acid (FA) stimulates neural stem cell (NSC) proliferation in vitro and enhances hippocampal neurogenesis in rats after middle cerebral artery occlusion (MCAO). The effect of FA supplementation on exogenous NSCs transplanted in MCAO rats was observed to determine if FA can stimulate NSC replacement after focal cerebral ischemia. Rats were randomly assigned to 3 groups: MCAO; MCAO and exogenous NSC transplantation (MCAO+NSCs); and MCAO, NSC transplantation and FA (MCAO+NSCs+FA). FA (0.8 mg/kg) or vehicle was administered by gavage daily for 28 days before MCAO and 23 days afterward. NSCs were labeled with superparamagnetic iron oxide (SPIO) and bromodeoxyuridine (BrdU) prior to transplantation into the striatum, contralateral to the ischemic zone, at 2 days post-MCAO. Magnetic resonance imaging tracking and fluorescent immunohistochemistry, as well as measurement of serum folate concentration, were performed at intervals up to 21 days after transplantation. FA supplementation caused sustained increases of 400-600% in serum folate concentration. Magnetic resonance images indicated that SPIO-labeled NSCs were more abundant at the transplantation and ischemic brain sites in MCAO+NSCs+FA rats than in MCAO+NSCs rats. Similarly, immunohistochemistry showed that the numbers of Sox-2/BrdU double positive cells at the transplantation and ischemic sites were higher in the rats that received FA. In conclusion, after focal cerebral ischemia, FA supplementation stimulates transplanted NSCs to proliferate and migrate to ischemic sites.


Brain Ischemia/physiopathology , Folic Acid/pharmacology , Neural Stem Cells/transplantation , Animals , Cell Movement , Cell Proliferation , Dextrans , Folic Acid/blood , Infarction, Middle Cerebral Artery/physiopathology , Magnetic Resonance Imaging , Magnetite Nanoparticles , Male , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation/methods
17.
Antioxid Redox Signal ; 19(17): 2129-40, 2013 Dec 10.
Article En | MEDLINE | ID: mdl-23682970

SIGNIFICANCE: Evidence is emerging that parenteral administration of high-dose vitamin C may warrant development as an adjuvant therapy for patients with sepsis. RECENT ADVANCES: Sepsis increases risk of death and disability, but its treatment consists only of supportive therapies because no specific therapy is available. The characteristics of severe sepsis include ascorbate (reduced vitamin C) depletion, excessive protein nitration in microvascular endothelial cells, and microvascular dysfunction composed of refractive vasodilation, endothelial barrier dysfunction, and disseminated intravascular coagulation. Parenteral administration of ascorbate prevents or even reverses these pathological changes and thereby decreases hypotension, edema, multiorgan failure, and death in animal models of sepsis. CRITICAL ISSUES: Dehydroascorbic acid appears to be as effective as ascorbate for protection against microvascular dysfunction, organ failure, and death when injected in sepsis models, but information about pharmacodynamics and safety in human subjects is only available for ascorbate. Although the plasma ascorbate concentration in critically ill and septic patients is normalized by repletion protocols that use high doses of parenteral ascorbate, and such doses are tolerated well by most healthy subjects, whether such large amounts of the vitamin trigger adverse effects in patients is uncertain. FUTURE DIRECTIONS: Further study of sepsis models may determine if high concentrations of ascorbate in interstitial fluid have pro-oxidant and bacteriostatic actions that also modify disease progression. However, the ascorbate depletion observed in septic patients receiving standard care and the therapeutic mechanisms established in models are sufficient evidence to support clinical trials of parenteral ascorbate as an adjuvant therapy for sepsis.


Ascorbic Acid/pharmacology , Sepsis/drug therapy , Animals , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Ascorbic Acid/therapeutic use , Blood Coagulation/drug effects , Chemotherapy, Adjuvant , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Microvessels/drug effects , Microvessels/physiopathology , Sepsis/mortality
18.
Cell Biochem Biophys ; 66(3): 559-66, 2013 Jul.
Article En | MEDLINE | ID: mdl-23292356

The present study investigated the roles of folic acid and DNA methyltransferases (DNMTs) in the differentiation of neural stem cells (NSCs). Neonatal rat NSCs were grown in suspended neurosphere cultures and identified by their expression of SOX2 protein and capacity for self-renewal. Then NSCs were assigned to five treatment groups for cell differentiation: control (folic acid-free differentiation medium), low folic acid (8 µg/mL), high folic acid (32 µg/mL), low folic acid and DNMT inhibitor zebularine (8 µg/mL folic acid and 150 nmol/mL zebularine), and high folic acid and zebularine (32 µg/mL folic acid and 150 nmol/mL zebularine). After 6 days of cell differentiation, immunocytochemistry and western blot analyses were performed to identify neurons by ß-tubulin III protein expression and astrocytes by GFAP expression. We observed that folic acid increased DNMT activity which may be regulated by the cellular S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH), and the abundance of neurons but decreased the number of astrocytes. Zebularine blocked these effects of folic acid. In conclusion, folic acid acts through elevation of DNMT activity to increase neuronal differentiation and decrease astrocytic differentiation in NSCs.


Cell Differentiation/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , Folic Acid/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurons/cytology , Animals , Cytidine/analogs & derivatives , Cytidine/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Neurons/drug effects , Rats , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism
19.
J Nutr Biochem ; 24(7): 1295-301, 2013 Jul.
Article En | MEDLINE | ID: mdl-23332600

The proliferative response of neural stem cells (NSCs) to folate may play a critical role in the development, function and repair of the central nervous system. It is important to determine the dose-dependent effects of folate in NSC cultures that are potential sources of transplantable cells for therapies for neurodegenerative diseases. To determine the optimal concentration and mechanism of action of folate for stimulation of NSC proliferation in vitro, NSCs were exposed to folic acid or 5-methyltetrahydrofolate (5-MTHF) (0-200 µmol/L) for 24, 48 or 72 h. Immunocytochemistry and methyl thiazolyl tetrazolium assay showed that the optimal concentration of folic acid for NSC proliferation was 20-40 µmol/L. Stimulation of NSC proliferation by folic acid was associated with DNA methyltransferase (DNMT) activation and was attenuated by the DNMT inhibitor zebularine, which implies that folate dose-dependently stimulates NSC proliferation through a DNMT-dependent mechanism. Based on these new findings and previously published evidence, we have identified a mechanism by which folate stimulates NSC growth.


Cell Proliferation/drug effects , DNA Modification Methylases/metabolism , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Animals , Dose-Response Relationship, Drug , Female , Folic Acid/administration & dosage , Neural Stem Cells/cytology , Pregnancy , Rats , Rats, Sprague-Dawley
20.
Cell Biochem Biophys ; 66(1): 131-7, 2013 May.
Article En | MEDLINE | ID: mdl-23109178

The aim of the present study was to determine if the excitatory amino acid homocysteine (Hcy) alters ERK signaling and cell proliferation in fetal neural stem cells (NSCs) in vitro. NSCs were isolated from fetal rats and grown in serum-free suspension medium. The cells were identified as NSCs by their expression of immunoreactive Sox2. NSCs were assigned to one of four treatment groups: vehicle control, low-dose Hcy group (Hcy-L, medium contained 30 µmol/L Hcy), middle-dose Hcy group (Hcy-M, 100 µmol/L Hcy) and high-dose Hcy group (Hcy-H, 300 µmol/L Hcy). Cell proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Protein expression levels of ERK1/2 and phosphorylated ERK1/2 were detected by Western blot. The effects of Hcy on NSC death, including apoptosis, were assessed by using flow cytometry and trypan blue exclusion. The results showed that NSCs grew as neurospheres in the serum-free medium. Hcy decreased ERK1/2 protein phosphorylation and NSC proliferation, but it did not induce cell death or apoptosis within the concentration from 30 to 300 µmol/L. The above results are consistent with the hypothesis that Hcy decreases fetal NSC proliferation by inhibiting ERK signaling.


Cell Proliferation/drug effects , Fetal Stem Cells/drug effects , Homocysteine/pharmacology , MAP Kinase Signaling System , Neural Stem Cells/drug effects , Animals , Apoptosis , Cell Count , Cell Survival/drug effects , Cells, Cultured , Culture Media, Serum-Free , Female , Fetal Stem Cells/cytology , Neural Stem Cells/cytology , Phosphorylation , Pregnancy , Rats , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
...